Oral Drug Delivery May 2011 Lo Res

download Oral Drug Delivery May 2011 Lo Res

of 40

Transcript of Oral Drug Delivery May 2011 Lo Res

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    1/40

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    2/40

    2

    CONTENTS

    The views and opinions expressed in this issue are those of the authors.

    Due care has been used in producing this publication, but the publisher

    makes no claim that it is free of error. Nor does the publisher acceptliability for the consequences of any decision or action taken

    (or not taken) as a result of any information contained in this publication.

    Front cover image: Multiple chrome pills, 3D4Medical.com/

    Science Photo Library.

    No Longer a Hit-or-Miss Proposition:

    Once-Daily Formulation for Drugs with

    pH-Dependent Solubility

    Gopi Venkatesh, Director of R&D & Anthony Recupero,

    Senior Director, Business Development

    Aptalis Pharmaceutical Technologies 4-8

    A possible approach for the desire to innovate

    Brian Wang, CEO & Dr Junsang Park, CSO

    GL PharmTech 10-13

    COMPANY PROFILE -

    Mayne Pharma International 14-15

    From Powder to Pill: A Rational Approach to

    Formulating for First-into-Man Studies

    Dr Robert Harris, Director, Early Development

    Molecular Profiles Ltd 16-19

    LiquiTime* Oral Liquid Controlled Release

    Camille Rivail, Business Development Analyst & Dr Jean

    Chatellier, Vice-President, Alliance Management

    Flamel Technologies SA 20-21

    Formulation Flexibility Broadens the Scope

    for Oral Thin Film Technology

    Martha Sloboda, Business Manager

    & Dr Scott Barnhart, Technical Director

    ARx, LLC 22-24

    Solumer Technology: a Viable Oral Dosage

    Form Option for BCS Class II Molecules

    Dr Robert Lee, Vice-President, Pharmaceutical

    Development & Dr Amir Zalcenstein, CEO

    SoluBest, Ltd 26-29

    Controlled Drug Release: Novel Time-Delayed

    Formulations and their Clinical Evaluation

    Dr Carol Thomson, Chief Operating Officer

    Drug Delivery International Ltd 30-32

    Liquid-Fill Hard Two-Piece Capsules:

    The Answer to Many Product Development Issues

    Mr Gary Norman, Product Development Manager

    Encap Drug Delivery 34-36

    Multi-Tip Tooling: A Guide

    Dale Natoli, Vice-President

    Natoli Engineering Company, Inc 38

    Oral Drug Delivery:

    Formulation Selection Approaches

    & Novel Delivery Technologies

    This edition is one in the ONdrugDelivery series of pub-

    lications from Frederick Furness Publishing. Each issuefocuses on a specific topic within the field of drug deliv-

    ery, and is supported by industry leaders in that field.

    EDITORIAL CALENDAR 2011:

    June: Injectable Drug Delivery (Devices Focus)

    July: Injectable Drug Delivery (Formulations Focus)

    September: Prefilled Syringes

    October: Oral Drug Delivery

    November: Pulmonary & Nasal Drug Delivery (OINDP)

    December: Delivering Biotherapeutics

    SUBSCRIPTIONS:

    To arrange your FREE subscription (pdf or print) to

    ONdrugDelivery, contact:

    Guy Furness, Publisher

    T: +44 (0) 1273 78 24 24

    E: [email protected]

    SPONSORSHIP/ADVERTISING:

    To feature your company in ONdrugDelivery, contact:

    Guy Furness, Publisher

    T: +44 (0) 1273 78 24 24

    E: [email protected]

    MAILING ADDRESS:Frederick Furness Publishing

    48, Albany Villas, Hove, East Sussex, BN3 2RW

    United Kingdom

    PRODUCTION/DESIGN:

    Mark Frost

    www.frostmark.co.uk

    Oral Drug Delivery: Formulation Selection Approaches

    & Novel Delivery Technologies is published by

    Frederick Furness Publishing.

    Copyright 2011 Frederick Furness Publishing.

    All rights reserved

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    3/40

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    4/40

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing4

    MEDICATION ADHERENCE

    It is estimated that 33-69% of all medication-

    related hospital admissions in the US are due

    to poor medication adherence, with a resultant

    cost of approximately US$100 billion a year.1-6

    Taking medications exactly as prescribed and

    following appropriate lifestyle recommenda-

    tions is highly beneficial and may reduce the

    impact of side effects.

    Practitioners should always assess adher-

    ence to therapy and may improve adherence by

    emphasising the value of a patients regimen,

    making the regimen as simple as possible, and

    customising the regimen to the patients life-

    style.7 Simple dosing (one pill, once daily) can

    help maximise adherence, particularly when

    combined with reinforcing visits / messages

    from healthcare practitioners, despite the fact

    that 10-40% of patients on simple regimens

    continue to have imperfect dosing adherence.8,9

    WHY ARENT ONCE-DAILY ORAL

    DOSAGE FORMS AVAILABLE FOR

    ALL DRUGS?

    As the orally administered pharmaceutical

    dosage form passes through the human gastro-

    intestinal (GI) tract, drug should be releasedfrom the dosage form and be available in solu-

    tion at or near the optimal site for drug absorp-

    tion to occur.10-12 The rate at which the drug is

    released from a dosage form and goes into solu-

    tion is important for the kinetics of drug absorp-

    tion. The dosage form and hence the pharma-

    ceutical ingredient (API) is subjected to varying

    pH levels during GI transit.13-16 Specifically, pH

    varies from a minimum of about 1.2 to a maxi-

    mum of around 7.4 (stomach pH: 1.2-2.5, which

    increases to 3.5-6.1 upon consumption of food;

    bile pH: 7.0-7.4; pH 5.0-6.0 in small intestine;

    and pH: 6 to 7 in the large intestine).

    GI fluid volume and agitation can vary sig-

    nificantly, which has substantial impact on drug

    dissolution and absorption.17 Moreover, transit

    time may vary significantly in individual parts

    of the GI tract, depending on individual size and

    prevailing local conditions.18

    Truly once-daily dosage forms of many weak-

    ly basic drugs are not commercially available.

    Several attempts have been made in the past

    at developing once-daily delivery systems of

    weakly basic drugs, such as carvedilol, ondanse-

    tron, and dipyridamole, with limited success.19-22

    This is largely because the absorption of a weakly

    basic drug is critically affected by its solubility

    and the required total daily dose. The ability to

    maintain these drugs in a soluble form as the drug

    passes through the GI tract throughout the day has

    been a substantial challenge for oral formulators.

    SOLUBILITY ENHANCEMENT BYORGANIC ACIDS

    The solubility-enhancing property of

    organic acids23 is exploited during the manu-

    In this article, Dr Gopi Venkatesh, Director of R&D, and Dr Anthony Recupero, Senior

    Director, Business Development, both of Aptalis Pharmaceutical Technologies (formerly

    Eurand), describe a specific application of Diffucaps technology, which allows the creation

    of once-daily oral formulations of weakly basic active pharmaceutical ingredients, previously

    extremely difficult to achieve, but with significant benefits to patient adherence.

    NO LONGER A HIT-OR-MISS PROPOSITION:

    ONCE-DAILY FORMULATION FOR DRUGSWITH PH-DEPENDENT SOLUBILITY

    Anthony Recupero, PhD

    Senior Director, BusinessDevelopmentT: +1 267 759 9346E: [email protected]

    Aptalis Pharmaceutical

    Technologies

    790 Township Line RoadSuite 250

    Yardley, PA 19067United States

    www.AptalisPharmaceutical

    Technologies.com

    Gopi Venkatesh, PhD

    Director of R&D

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    5/40

    Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com

    facture of customised-release (CR) dosage

    forms using Diffucaps technology. The

    potential for in situ formation of acid addition

    compounds24 is averted by using a sustained-

    release (SR) coating membrane between the

    inner organic acid layer and the weakly basic

    drug layer. The SR-coating membrane thus

    applied, precisely controls the release of the

    organic acid ensuring drug is not retained in

    the dosage form for lack of solubilising acid

    in theDiffucaps formulation.

    DIFFUCAPS TECHNOLOGY

    Diffucaps technology in its simplistic

    form (see Schematic of the Time Pulsatile

    Release / Time Sustained Release (TPR/TSR)

    bead shown in Figure 1) involves the prepara-

    tion of:

    (1) drug-containing cores by drug-layering on

    inert particles

    (2) customised release (CR) beads by coating

    immediate release (IR) particles with one or

    more functional dissolution rate controlling

    polymers or waxes

    (3) combining one or more functional polymer

    coated Diffucaps bead populations into

    hard gelatin or hydroxypropyl methylcel-

    lulose (HPMC) capsules.24

    MECHANISM OF DRUG RELEASE

    FROM TPR/TSR BEADS

    The water-insoluble and enteric polymers

    are dissolved in a common solvent mixture

    and the solution is sprayed onto drug particles.These two polymers may exist as molecularly

    dispersed or as molecular clusters in the lag-

    time coating membrane applied on the drug

    cores (Figure 1).

    During dissolution testing in two-stage dis-

    solution media (first two-hour dissolution test-

    ing in 700 mL of 0.1N HCl and thereafter

    testing in 900 mL of pH 6.8 buffer obtained

    by adding 200 mL pH modifier) or upon oral

    administration, water or body fluid is blocked

    from imbibing into the core as the polymeric

    system is impermeable in the acidic medium

    or gastric fluid. When the pH of the medium

    is changed to 6.8 or following exit from the

    stomach, the penetrating dissolution medium or

    intestinal fluid selectively dissolves the enteric

    polymer molecules or molecular clusters start-

    ing from the outermost membrane layer, thereby

    creating tortuous nanopore channels for dis-

    solved drug to pass through.23

    The tortuosity increases with increasing

    coating thickness and/or decreasing enteric

    polymer content, and consequently, the drug

    release from the TPR beads having no barrier

    coat becomes sustained with increasing thick-

    ness of the TPR coating.

    DEVELOPMENT OF ONCE-DAILY

    DOSAGE FORMS OF WEAKLY

    BASIC DRUGS

    Below is shown the method for the prepara-

    tion25 of CR drug delivery systems comprising

    one or more IR, SR, TPR/TSR, Delayed-Release

    (DR) bead populations, themselves containing

    a weakly basic, nitrogen moiety-containing API

    such as ondansetron, carvedilol, dypiramidole,

    lamotrigine or iloperidone, which is moderately

    soluble at pH 6, and at least one pharmaceutically accept-

    able organic acid as a solubiliser (see the schemat-

    ics of SR organic acid bead & TPR/TSR bead

    containing a weakly basic drug shown in Figure 2).

    The method comprises the following steps:

    a) layering an organic acid on 25-30 mesh sugar

    spheres;

    b) applying an SR coating on acid-layered beads

    with a water-insoluble polymer to control the

    rate of release of the acid;

    5

    Figure 1: Diffucaps Customised Drug Release Bead (A) soaked in pH 1.2 or resident in the stomach and (B) soaked in pH 6.8 orin transit in the intestinal tract.

    Figure 2: Diffucaps: Customised Drug Release Bead for pH-sensitive Drugs(e.g. Ondansetron HCl).

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    6/40

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing6

    c) preparing IR beads by layering the weakly

    basic nitrogen moiety-containing API and

    applying a protective seal-coat with a water-

    soluble polymer;

    d) preparing SR beads by applying a barrier

    (SR) coating of a water-insoluble polymer

    on the IR beads to sustain the drug release

    over several hours (if needed);

    e) preparing TPR beads by applying a lag-time

    coating on IR beads or SR beads (called

    TSR beads) comprising water-insoluble and

    enterosoluble polymers for a weight gain

    sufficient to achieve a lag time (a time period

    of less than 10% drug release) of 2-6 hours

    followed by a sustained-release profile; and

    f) filling into a capsule a mixture of IR beads

    and one or more TPR bead populations at

    appropriate amounts to achieve a target phar-

    macokinetics profile suitable for a once-daily

    dosing regimen.

    The following examples demonstrate how

    Aptalis Pharmaceutical Technologies utilised

    the above process to formulate once-daily dos-

    age forms of ondansetron and iloperidone.

    NAUSEA AND VOMITING

    FOLLOWING CHEMOTHERAPY,

    RADIATION THERAPY, OR SURGERY

    Radiotherapy-induced nausea and vomit-

    ing (RINV), chemotherapy-induced nausea and

    vomiting (CINV), and postoperative nausea and

    vomiting (PONV) remain the most common anddistressing challenges facing patients receiving

    these cancer therapies or following surgical pro-

    cedures under general anaesthesia (occurring in

    up to 80% of cases).25-33

    Nausea and vomiting very often occur together

    but can also occur independently. RINV and

    CINV during cancer therapy can have a direct

    and significant impact on adherence to primary

    therapy. Some of the most highly prescribed anti-

    emetics suffer from a short-half life requiring mul-

    tiple daily doses for control of emesis. Between

    doses, the plasma levels of the anti-emetic can

    drop well below efficacious levels increasing

    the risk for breakthrough nausea and vomiting,

    particularly when subsequent doses are not taken

    exactly as scheduled. Proper control of acute and

    breakthrough nausea and vomiting therefore can

    be achieved with a higher probability and a higher

    level of confidence with a customised-release

    (CR) dosage form for oral administration, prefer-

    ably administered prior to the procedure.

    Weakly basic ondansetron HCl dihydrate

    Ondansetron HCl dihydrate, the API in the

    branded product, Zofran Tablets (4 and 8 mg

    base equivalent) and Zofran Oral Solution,

    marketed by GlaxoSmithKline, is a selective

    serotonin 5-HT3

    blocking agent (an antiemetic).

    The API in Zofran ODTs (orally disintegrating

    tablets, 4 and 8 mg) is ondansetron base. All

    products are immediate release (IR) formulations.

    Ondansetron is indicated for the prevention of

    nausea and vomiting associated with radiotherapy

    (adults: 8 mg tid) and/or chemotherapy (adults:

    8 mg bid to tid) and prevention of postoperative

    nausea and/or vomiting (adults: 8 mg bid).

    Ondansetron is a weakly basic drug having a

    pKa of 7.4 and an elimination half-life averag-ing approximately 3.81 hours. It is practically

    insoluble in the pH environment of the intestinal

    tract. However, there is a dramatic increase

    in solubility in aqueous organic acid solution,

    making it a good candidate for developing once-

    daily dosage forms based on the organic acid

    approach ofDiffucaps technology.

    Modified release (MR), once-daily dosage

    forms of ondansetron HCl dihydrate using

    Diffucaps technology

    Pharmacokinetic/biopharmaceutical modeling

    and simulation of possible plasma profiles based

    on available pharmacokinetic data as a guide

    in the design of customised-release (CR) dos-

    age forms in order to be suitable for a once-

    daily dosing regimen is typically performed using

    WinNonlin and/or GastroPlus computer simu-

    lation and modeling techniques. The CR capsule

    product was designed to comprise appropriate

    amounts of both IR and TPR components wherein

    the TPR component used SR-coated organic acid

    beads as inert cores to design multiple TPR beadpopulations with different lag times.33 The use

    of such methods resulted in reduced feasibility

    development time and enhanced the probability

    of success of the program.

    For the IR component of the formulation,

    rapid release (RR) granules comprising ondan-

    setron, mannitol, and organic acid were devel-

    oped, which are designed to release the drug

    faster than, or similar to,Zofran IR tablets even

    under alkaline pH conditions.33

    Ondansetron HCl CR capsules were

    designed to comprise appropriate amounts of

    both RR granules and TPR beads. Three CR

    formulations were prepared for pharmacokinetic

    (PK) testing in healthy volunteers.33

    A randomised, four-way crossover pilot PK

    study was conducted that included 12 healthy

    male volunteers, aged 18-55 years, with a wash-

    out period of seven days. Each volunteer was

    dosed with one of three test formulations of

    Ondansetron MR at 0800h, or two Zofran (8

    mg) at 0800h and 1630h after an overnight fast.

    Figure 3 shows the mean plasma concentration-

    time profiles achieved. The relative bioavail-

    ability compared with 8 mg IR bid reference

    was approximately 0.85 for all test formulations

    (Test Formula 1, 2, and 3) at the end of 24 hours.

    Based on these results, Test Formula 3, given

    the product code EUR1025, was advanced into

    pivotal PK studies which have been completed.34

    In these trials, single and repeated oral adminis-

    trations of 24 mg EUR1025 resulted in similar

    rate and extent of exposure as 8 mg Zofran tid.

    Steady-state concentrations of Treatment 2 (8 mg

    Zofran bid) and Treatment 3 (8 mgZofran tid)

    are equivalent to that of single administrations of

    two and three 8 mg Zofran

    , respectively.34

    Thetotal exposure of ondansetron (AUC

    0-24) from

    EUR1025 on day six was approximately 13%

    higher than that observed on day one, suggesting

    minor accumulation following repeated dosing.

    Figure 3: Pilot PK Study - Ondansetron QD versus Ondansetron IR (Zofran).

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    7/40

    Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com 7

    The total exposure of Treatment 1 (24 mg

    EUR 1025) appears to be nearly equivalent to

    that of Treatment 3 (8 mgZofran tid) at steady

    state. The product is now ready to enter Phase III

    clinical development.34

    ILOPERIDONE TPR BEADS AND

    RELEASE PROFILES

    The Diffucaps organic acid approach used

    with ondansetron is applicable to any weakly

    basic drug, which is at least slightly soluble at a

    pH3, but is poorly soluble or practically insolu-

    ble above pH 6. Iloperidone, the API in Fanapt,

    is a weakly basic, dopamine and serotonin recep-

    tor antagonist exhibiting antipsychotic activities.

    Iloperidone (12 mg) is dosed twice daily.

    The incidence of adverse effects in

    patients treated with Fanapt 20-24 mg/daywere twice that occurring in patients treated

    with Fanapt 10-16 mg/day indicating an MR,

    once-daily formulation may improve the side

    effect profiles of iloperidone. Initial studies

    indicate that by combining IR and TPR bead

    populations at appropriate quantities (as deter-

    mined by simulation and modeling) to provide

    desired in vitro release profiles, it would be

    possible to achieve target plasma profiles suit-

    able for a once-daily dosing regimen.

    ADVANTAGES OF CR DIFFUCAPS

    DRUG DELIVERY SYSTEMS

    Controlled-release drug delivery systems

    consisting of coated multiparticulates, particu-

    larly based on Diffucaps technology, which

    typically have a particle size in the range of 200-

    600 m, exhibit characteristic target in vitro

    profiles, as well as target plasma concentration-

    time profiles to be suitable for a once-daily

    dosing regimen.

    Multiparticulate drug delivery systems, such

    as Diffucaps, offer the following advantages

    over conventional controlled-release monolithic

    dosage forms such as matrix or coated tablets

    including osmotic delivery systems:

    Dispersed along the GI Tract for more effec-

    tive delivery

    Predictable and consistent GI transit time

    thereby minimising food effect

    Low probability of dose dumping

    Reduced inter- and intra-subjectvariability

    Easy adjustment of multiple dose strengths

    In addition, the Diffucaps technology offers

    incremental advantages: Easy adjustment of target plasma profiles

    including combining bead populations exhibit-

    ing differing release profiles

    Ability to create combination products of

    incompatible actives or actives requiring dif-

    fering target plasma profiles

    Capability to create micro-environments:

    Create a sustainable acidic pH micro-envi-

    ronment within coated bead to solubilise

    the weakly basic drug (which is practically

    insoluble at pH 6.0 or above) in order to

    extend its release into the GI tract

    Create a sustainable alkaline pH micro-

    environment within coated bead to moder-

    ate the solubility of a weakly basic drug

    (which is extremely soluble in the entire

    physiologically relevant pH range of 1.0 to

    8.0) to avoid dose dumping

    Improve patient adherence due to reduced fre-

    quency of dosing, ease of oral administration,

    reduction in incidence of adverse events, and/

    or improved safety profile

    Additional product patent protection

    CONCLUSIONS

    Adherence to oral medication regimens,

    and therefore effective therapy, is a common

    issue for patients across multiple indications.

    Although simple dosing regimens (one pill,

    once daily) as provided by extended release

    (ER) formulations for a number of products

    are available, there are still many drugs for

    which an ER, once-daily form has proven to

    be exceptionally challenging to develop. These

    challenging molecules frequently have water

    solubility issues which may also be complicated

    by limited molecule half-life.

    The Diffucaps technology is one approach

    that effectively overcomes such challenges,

    allowing for straightforward development of

    ER, once-daily formulations that help to improve

    adherence, which can result in improved effi-

    cacy and patient quality of life.

    REFERENCES

    1. McDonnell PJ, Jacobs MR. Hospital admis-

    sions resulting from preventable adverse reac-

    tions. Ann Pharmacother 2002; 36:1331-6.

    2. Senst BL, Achusim LE, Genest RP, et al.

    Practical approach to determining costs

    and frequency of adverse drug events in a

    healthcare network. Am J Health Syst Pharm

    2001; 58:1126-32.

    3. Levy G, Zamacona MK, Jusko WJ. Developing

    compliance instructions for drug labeling.

    Clin Pharmacol Ther 2000;68:586-91.

    4. Berg JS, Dischler J, Wagner DJ, Raia JJ,

    Palmer-Shevlin N. Medication compliance:a healthcare problem. Ann Pharmacother

    1993;27:Suppl 9:S1-S24.

    5. LaRosa JC. Poor compliance: the hidden risk

    factor. Curr Atheroscler Rep 2000;2: 1-4.

    6. Horwitz RI, Horwitz SM. Adherence to treat-

    ment and health outcomes. Arch Intern Med

    1993;153:1863-8.

    7. Osterberg, L, and Blaschke, T. Adherence to

    Medication. N Engl J Med 2005; 353:487-97.

    8. Greenberg RN. Overview of patient compli-

    ance with medication dosing: a literature

    review. Clin Ther 1984;6:592-9.

    9. Eisen SA, Miller DK, Woodward RS,

    Spitznagel E, Przybeck TR. The effect of

    prescribed daily dose frequency on patient

    medication compliance. Arch Intern Med

    1990;150:1881-4.

    10. P.K. Gupta, J.R. Robinson, Oral controlled

    release delivery, in: Treatise on controlled

    drug delivery, Kydonieus, A. (ed.), Marcel

    Dekker, New Jersey (1992) 255-310.

    11. R.E. Notari, Biopharmaceutics and clini-

    cal pharmacokinetics, Marcel Dekker,Inc., New York, 1987.

    12. D.J. Greenblatt, L.L. van Moltke,

    J.J. Harmatz, and R.I. Shader,

    Pharmacokinetics, pharmacody-

    namics, and drug disposition, In:

    Neuropsychopharmacology, K.L Davis,

    D. Charney, J.T. Coyle, and C. Nemerof

    (eds), Lippincott, Williams & Wilkins,

    Philadelphia, (2002) 507-524.

    13. G. Chawla, P. Gupta, V. Koradia, and

    A.K. Bansal, A means to address regional

    variability in intestinal drug absorption,

    Pharm. Technol., July 2003, 50-68.

    14. Davis, S.S., 1987. Evaluation of the gas-

    trointestinal transit and release charac-

    teristics of drugs. in. Johnson, P., Lloyd-

    Jones, J.G. (Eds), Drug Delivery systems

    [Fundamentals and techniques]. Ellis

    Harwood, Chichester, pp. 164-179.

    15. J.B. Dressman, R.R. Berardi, L.C.

    Dermentzoglou, T.L. Russell, S.P.

    Schmaltz, J.L. Barnett, K.M. Jarvenpaa,

    Upper gastrointestinal (GI) pH in young

    healthy men and women, Pharm. Res. 7

    (1990) 756761.

    16. C. Schiller, C.P. Prohlicht, T. Giessmann,

    W.,Siegmund, H. Monnikess, and N.

    Hosten, Intestinal fluid volume and transit

    of dosage forms as assessed by magnetic

    resonance imaging, Aliment. Pharmacol.

    Ther., 22 (2005) 871-879.

    17. Davis, S.S., Hardy, J.G., Taylor, M.J.,

    Whalley, D.R., Wilson, C.G., 1984. A com-

    parative study of the gastrointestinal transit

    of a pellet and tablet formulation. Int. J.

    Pharm. 21, 167-177.

    18. W. Eisert and P. Gruber, US 6,015,577B1: Pharmaceutical compositions con-

    taining dipyridamole or mopidamol and

    acetylsalicylic acid or the physiologically

    acceptable salts thereof; processes for

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    8/40

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing8

    preparing them and their use in treat-

    ing clot formation, assigned to Dr. Karl

    Thomae GmbH.

    19. G.K. Jain, O. Anand, and A. Rampal, WO

    2004/096182 A1: Extended release matrix

    tablets of carvedilol, assigned to Ranbaxy

    Laboratories Limited.

    20. V. Andronis, K. A. Lamey, and C.K. Oh,

    US 20040019096 A1: Novel formulations

    of carvedilol, assigned to SmithKline

    Beecham Corporation.

    21. February 6, 2007 at 3:00 PM: (Business

    Wire) Scolr Pharma announces positive

    results from its Second 24 Hour CDT

    Ondansetron Trial at http://www.scolr.com.

    22. G. Venkatesh, US 20070196491 A1: Drug

    delivery systems comprising weakly basic

    drugs and organic acids, assigned to

    Eurand, Inc.23. G. Venkatesh, Diffucaps technology

    for controlled release drug delivery, In.

    Chronotherapeutics, B.-B.C.Youan (Ed.),

    John Wiley & Sons, New York (2009)

    121-144.

    24. R. Sun, K.W. Klein, and P.F. White,

    The effect of timing of ondansetron

    administration in outpatients undergoing

    Otolaryngologic surgery. Anesth. Analg.

    84 (1997) 331-6.

    25. J.L. Parlow, A.T. Meikle, J. v. Vlymen,

    Post discharge nausea and vomiting after

    ambulatory laparoscopy is not reduced by

    promethazine prophylaxis, Can. J. Anesth.

    46 (1999) 719-724.

    26. T.J. Gan, R. Franiak, J. Reeves,

    Ondansetron orally disintegrating tablet

    versus placebo for the prevention of post-

    discharge nausea and vomiting after ambu-

    latory surgery, Anesth. Analg. 94 (2002)

    1199-1200.

    27. F. Roila, P.J. Hesketh, J. Herrstedt,

    Antiemetic Subcommittee of the

    Multinational Association of Supportive

    Care in Cancer. Prevention of chemo-

    therapy and radiotherapy induced emesis:

    results of the 2004 Perugia International

    Antiemetic Consensus Conference, Ann.

    Oncol. 17 (2006) 20-28.28. T.J. Gan. Risk factors for postoperative

    nausea and vomiting Anesth. Analg. 102

    (2006) 1884-98.

    29. D.S. Wagner, V. Gauger, D. Chiravuri,

    and K. Faust, Ondansetron oral disinte-

    grating tablet for the prevention of post-

    operative vomiting in children undergoing

    strabismus surgery, Ther. Clin. Risk

    Manag. 3 (2007) 691-694.

    30. T.J. Gan, C.C. Apfel, A. Kovac, B.K. Philip,

    N. Singla, H. Minkowitz, A.S. Habib, J.

    Knighton, A.D. Carides, H. Zhang, K.J.

    Horgan, J.K. Evans, F.C. Lawson, and

    The Aprepitant-PONV Study Group, A

    randomzed, double-blind comparison of the

    NK1 antagonist, Aprepitant, versus ondan-

    setron for the prevention of postoperative

    nausea and vomiting, Anesth. Analg. 104

    (2007) 1082-1089.

    31. P.C. Feyer, E. Maranzano, A.M.

    Molassiotis, F. Roila, R.A. Clark-Snow,

    and K. Jordan, Radiotherapy induced nau-

    sea and vomiting (RINV): MASCC/ESMO

    guideline for antiemetics in radiotherapy:

    Update 2009, Support Care Cancer

    Published online: 10 August 2010, DOI

    10.1007/s00520-010-0950-6.

    32. B. Nevidjon and R. Chaudhary, Controlling

    emesis: Evolving challenges, novel strate-gies, The J. Support. Oncol. 8 (2010) 1-10.

    33. G. Venkatesh, J.-W. Lai, N.H. Vyas, and V.

    Purohit, US 20090232885 A1: Drug deliv-

    ery systems comprising weakly basic drugs

    and organic acids, assigned to Eurand, Inc.

    34. G. Venkatesh, S. Perrett, and R. Thieroff-

    Ekerdt, US 20090232885 A1: Methods

    of treating PDNV and PONV with ER

    Ondansetron compositions, assigned to

    Eurand, Inc.

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    9/40

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    10/40

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing10

    How did you feel when you heard your brand

    product was easily copied by a generic company

    after the expiration of its new chemical entity

    patent? And what about the case when someone

    from sales & marketing came and complained

    of setbacks in developing a pre-defined refor-

    mulation product?...

    For various reasons, with which readers

    will already be familiar, individuals working

    in pharmaceutical product development and

    formulation have been under significant pres-

    sure for some time. This pressure may have

    made possible various kinds of open-innovation

    by prompting the adoption of technologies or

    products from outside.

    The drug delivery industry has been work-

    ing as an innovator and excellent partner over

    the past 30 years, providing technologies that

    have enabled brand pharmaceutical compa-

    nies to take new steps. This is surely one rea-

    son why the number of reformulated productsreached about triple that of new chemical

    entities (NCEs) in 2009 (75 versus 26).1 As

    a player in the oral drug delivery field, we at

    GL PharmTech were pleased to note that oral

    drug delivery products captured about 10% of

    the top 200 product sales, which reportedly

    reached US$14.5 billion.

    UNDER PRESSURE FOR

    REFORMULATION

    As product developers using oral drug deliv-

    ery technology, GL PharmTech is constantly

    considering what gaps innovators want to fill in

    their currently marketed products. What should

    be the factor to drive reformulation?

    There are many reasons why currently mar-

    keted products could be reformulated. These

    can originate from aspects of marketing, manu-

    facturing, regulation, generic competition, and

    even sometimes a purely scien-

    tific basis. These various rea-

    sons can come alone, together,

    or complicatedly combined.

    Therefore, a single outside

    technology or reformulated

    product could not fill all the gaps

    or cover possible voids the inno-

    vator did not feel compelled to

    address at one time. This might

    be the driving force for why

    innovative pharma companies

    have their departments of devel-

    opment review outside technology as often as

    possible and compile it in their databases.

    Whenever we imagine someone at an inno-

    vator company trying to align all the variables

    to find a fit for their molecules or productswith outside drug delivery technologies, the

    picture gives a strong feeling that a new drug

    delivery player might be what is required to

    make every thing click together.

    Here, Hunsik (Brian) Wang, Chief Executive Officer, and Junsang Park, PhD, Chief ScientificOfficer, both of GL PharmTech, introduce GLARS, a novel concept extended-release triple-

    layered tablet delivery technology for delivery to the intestine and colon.

    A POSSIBLE APPROACH FOR THE DESIRETO INNOVATE

    Hunsik (Brian) Wang

    Chief Executive OfficerT: +82 31 739 5220 (Ext. 102)F: +82 31 739 5034E: [email protected]

    Dr Junsang Park

    Chief Scientific OfficerT: +82 31 739 5220 (extension 301)F: +82 31 739 5220E: [email protected]

    GL PharmTech138-6 Sangdaewon JungwonSeongnamRepublic of Korea (South Korea)

    HOW DID YOU FEEL

    WHEN YOU HEARD YOUR BRAND

    PRODUCT WAS EASILY COPIED

    BY A GENERIC COMPANY AFTER

    THE EXPIRATION OF ITS NEW

    CHEMICAL ENTITY PATENT?

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    11/40

    Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com 11

    NEEDS FOR MEETING A NEW

    CONCEPT IN ORAL EXTENDED

    RELEASE

    This situation could be particularly true in

    the field of oral extended-release dosage forms.

    The first big successes OROS from Alza(now Johnson & Johnson, New Brunswick,

    NJ, US) and Geomatrix from Skyepharma

    (London, UK) had a large impact in the field

    of oral extended-release drug delivery technol-

    ogy. However, there has not since been a other

    strong player showing a comparable, remark-

    able success, and the platform patents of both

    technologies have expired. In addition, the rela-

    tively short gastro-intestinal transit time cannot

    expectedly or unexpectedly give a new start

    to blockbuster products, even by applying the

    already-existing technologies. In other words,

    the molecule candidates on the market or under

    development must have a suitable half-life for

    those technologies to be applied.

    Recently, a novel oral extended release tech-

    nology was presented. Astellas Pharma (Tokyo,

    Japan; formerly Yamanouchi Pharma) suggest-

    ed a possible cause for limited absorption in the

    colon and developed a new dosage form capable

    of dragging and retaining gastro-intestinal fluid

    into the dosage form itself, which could, in turn,

    act as drug-releasing media in the colon.3, 4

    They found another main reason for mal-

    absorption in the colon to be that there was

    no additional surrounding fluid present for

    active substance in dosage form to be released

    from, and described how this limitation could

    be overcome to some degree by incorporating

    highly water-retaining polymers into the dosage

    form. They named this technology OCAS (Oral

    Controlled Absorption System).

    Up until now, Astellas has applied this tech-

    nology to at least two products, according to the

    literature, including tamsulosin, a global lead-

    ing drug for anti-benign prostatic hyperplasia

    (BPH), and mirabegron, an anti-incontinencedrug. The reformulated tamsulosin product has

    been on sale in European regions under various

    local brand names such as Alna OCAS, Omnic

    OCAS, Flomaxtra XL, Urolosin OCAS and

    Praf T. Mirabegron has been in Phase III clini-

    cal trials in various countries.

    The reformulated OCAS tamsulosin product

    was reported to show not only higher night-time

    maintenance of plasma concentrations during

    but also no food effects upon its pharmacoki-

    netic profiles.5, 6

    GLARS: A NOVEL INTESTINAL AND

    COLONIC EXTENDEND-RELEASE

    TECHNOLOGY

    The focus of GL PharmTech over the past

    ten years has been on developing a technol-

    ogy named GLARS (Geometrically Long

    Absorption Regulated System). The system

    entraps more gastro-intestinal fluid into the dos-

    age form at early dissolution time to give further

    extended absorption in the colon.

    We have now reached a remarkable milestone.

    During the course of our work, we fabricated a

    triple-layered tablet, where the drug and very

    hydrophilic excipients are incorporated into the

    middle layer while highly water-retaining and

    swellable materials are embedded in the upper

    and lower layers (see Figure 1).

    After oral administration, the surrounding

    GI fluid can penetrate very quickly into the

    middle layer, thus the upper and lower layers

    concurrently swell rapidly. These rapidly swol-

    len upper and lower layers enclose the lateral

    side of the middle layer in quick-time (as shown

    in Figure 2).

    The amount of water drawn into the tablet

    reaches about 3-5 times the weight of the tablet

    itself and it can function, in turn, as additionalmedia which enables further later drug release out

    of the dosage form when it passes into the colon.7

    The key feature of GLARS is the middle

    layer, where it horizontally divides the tablet

    structure. As long as the surrounding water

    penetrates into the tablet core, it can perform its

    role to diffuse outward from the core. During

    the diffusion process the water can also move

    upwards and downwards, and this additional

    diffusion, together with the diffusion of GI fluid

    present outside the tablet, allows the upper and

    lower layers to be quickly swollen and gelled,

    at the same time.

    As is already recognised in the field, a

    conventional matrix sustained-release tablet

    has its own erosion, diffusion, swelling front,

    and un-swollen intact core. Achieving com-

    plete swelling of a tablet without an intact

    core before considerable erosion during normal

    gastro-intestinal transit time has appeared to be

    challenging. From this standpoint the insertion

    of a highly water-penetrating middle layer into

    GLARS was a radical approach.

    Another feature of this system is rapid

    enclosing of the tablets lateral side with the

    upper and lower layers in a relatively short time.

    As shown in Figure 3, after closing, drug release

    is mainly demonstrated through the enclosed

    lateral side, where the orange colour (from the

    incorporated colourant) in the middle layer is

    much thicker than on the other sides.

    PROOF OF CONCEPT

    Tamsulosin

    The first target for determining whether this

    system could actually operate was the block-

    buster molecule, tamsulosin.8 Marketed under

    the name Harnal, as well as Flomax, this

    product was originally formulated into enteric-matrix granules in a hard gelatin capsule. In

    Asia, including Japan and Korea, a normal

    dose is 0.2 mg, compared with 0.4 mg in the

    Americas and Europe.

    Figure 1: Triple-layered structure ofGLARS

    Figure 2: Morphological changes in GLARS upon water contact

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    12/40

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing12

    As presented in Figure 4, Tamsulosin

    GLARS, including a double amount of the API

    (0.4mg), showed a nearly similar peak con-

    centration to Harnal containing only 0.2 mg

    of the API. Nonetheless, the extent of absorp-

    tion, AUC, was not reduced but, instead, nearly

    doubled.

    When considering normal cases of most

    types of drug product with dose proportionality

    the greater the dose administered, the propor-

    tionally higher the pharmacokinetic parameters

    Cmax

    and AUC. However, the GLARS system

    demonstrated a proportionally higher extent of

    absorption without a remarkable increase in the

    rate of absorption. This result suggests that the

    system can be applied to types of drugs with

    the very close relationship of peak concentra-

    tion versus adverse effects, for which extended

    release dosage forms are desired.

    Another finding in the application was that

    the therapeutic concentration was persistent

    even during the night. Considering reports that

    nocturia is a key worry frequently raised by

    BPH patients, longer duration of action at night

    could be a very meaningful step for meeting

    patients ongoing needs.9

    The relatively rigid swollen matrix structure

    of GLARS formulations allows drug release to

    be unaffected by surrounding mechanical flux,

    which can provide relatively consistent in vivo

    drug release irrespective of the degree of gastro-

    intestinal motility.

    Tianeptine

    Another proof on concept study was car-

    ried out with tianeptine, an anti-depressant,

    developed and marketed under the name

    Stablon by Servier (Neuilly-sur-Seine,

    France). The purpose of the application was

    to determine whether the system could reduce

    the number of daily administrations for better

    patient compliance.

    Figure 5 represents the results of the pharma-

    cokinetic study, where the total amount of the

    API was the same, 37.5 mg. In terms of the phar-

    macokinetic parameters, no large difference was

    shown between Tianeptine GLARS (GX-2903)

    once daily, and three-times-daily administration

    of the immediate-release dosage form.

    Of course, this should be further evaluated

    to determine whether this kind of plasma profile

    is clinically effective and comparable with theperformance of existing immediate-release dos-

    age forms.

    CREATING EARLY PARTNERSHIPS

    Several oral drug delivery technologies have

    come and gone, and new systems still emerge

    even today. However, their fates appear to be

    very similar to those of NCEs. Approximately

    five years is needed to demonstrate any phar-

    maceutical or clinical evidence of one technol-

    ogy. In addition, reformulated products must be

    exclusively marketed for at least ten years.

    Then, we, as drug delivery industry workers,

    have only five years between showing evidence

    and launching a product into market.

    Another aspect to be considered is that

    there comes a time when additional innovative

    pharmaceutical applications are needed over the

    previously much-used simple matrix-type sus-

    tained release form. When exclusivity expires,

    there is the likely tendency of copying by

    generic companies in a very short time.

    Considering both aspects in combination,

    the marriage of the NCE with the drug delivery

    system, through a partnership between pharma

    company and drug delivery company, should be

    created as early as possible.

    Early partnering would represent a great step

    towards securing more valuable next-generation

    reformulated products.

    REFERENCES:

    1. Rekhi GS. Advances in solid dose oral

    drug delivery. ONdrugDelivery: Oral drug

    Delivery & Advanced Excipients, 2010, 14-18.2. Bossart J .Oral drug delivery:

    the numbers behind the business.

    ONdrugDelivery: Oral drug Delivery &

    Advanced Excipients, 2010, 4-6.

    Figure 4: Pharmacokinetic profiles of Tamsulosin GLARS, which shows doubled extentof absorption without a dose-proportional increase of peak concentration

    Product Cmax (ng/mL) AUCt (hr ng/mL

    Harnal (0.2mg; qd) 5.160.97 69.622.3

    GLPTs GLARS (0.4mg; qd 6.602.70 114.3439.9

    Figure 3: Schematic representation of rapid water penetration through middle layer aswell as swelling and enclosing of upper and lower layers

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    13/40

    Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com

    3. Sako K et al. Influence of physical fac-

    tors in gastrointestinal tract on acetami-

    nophen release from controlled-release

    tablets in fasted dogs. Proceedings of

    the 6th Conference of the Academy of

    Pharmaceutical Science and Technology,

    Japan. 1990, 30-31.

    4. Sako K et al. Relationship between gela-

    tion rate of control-release acetaminophen

    tablets containing polyethylene oxide and

    colonic drug release in dogs. Pharm Res,

    1996, 13(4), 594-598.

    5. Michel MC et al. The pharmacokinetic pro-

    file of Tamsulosin oral controlled absorption

    system(OCAS). Eur. Urol. Suppl. 2005,

    4, 15-24

    6. Djavan B et al. The impact of Tamsulosin

    oral controlled absorption system(OCAS)

    on nocturia and the quality of sleep: pre-

    liminary results of a pilot study. Eur Urol

    Suppl, 2005, 4, 61-68.

    7. Park JS et al. A novel three-layered tab-

    let for extended release with various layer

    formulations and in vitro release profiles.

    Drug Devel Ind Pharm, 2011, 37 (in press).

    8. Park JS et al. Formulation variation and

    in vitro-in vivo correlation for a rapidlyswellable three-layered tablet of Tamsulosin

    HCl. Chem Pharm Bull, 2011, 59 (in

    press).

    9. Schulman CC et al. The impact of noc-

    turia on health status and quality of

    life in patients with lower urinary tract

    symptoms suggestive of benign prostatic

    hyperplasia(LUTS/BPH). Eur Urol Suppl,

    2005, 4, 1-8.

    13

    Figure 5: Pharmacokinetic profiles of Tianeptine GLARS, which shows the possibility ofonce daily administration

    Product Cmax (ng/mL) AUCt (hr ng/mL

    Stablon (12.5mg x tid) 335107.6 2705.3601.8

    GLPTs GLARS (37.5mg qd 359.274.2 2849.7622.9

    ONdrugDelivery is now fi rmly established worldwide.

    It is the leading sponsored themed drug delivery publication.

    www.ondrugdelivery.com

    WEKNOWDRUG DELIVERYWant to KNOWdrug delivery too?Just subscribe FREE to ONdrugDelivery online today!

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    14/40

    14 www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing

    A leading pharmaceutical organisation built

    on a heritage of 160 years of industry excel-

    lence, Mayne Pharma International is a

    market-driven company offering a range of

    drug delivery technologies. Mayne Pharma

    International offers contract development

    and commercial manufacture for oral andtopical pharmaceutical products.

    Mayne Pharma International has comprehen-

    sive experience in the solid oral Drug Delivery

    System (DDS) market, encompassing develop-

    ment and manufacture of these products.

    The company has:

    more than 30 years experience in suc-

    cessfully developing DDS products for the

    global market a dedicated product development facility

    which meets cGMP standards, and includes

    pilot-scale plant equipment; this allows a

    scale-up pathway from small clinical trial

    batches to full commercial manufacture

    proven ability to develop and successfully

    transfer manufactured product and technol-

    ogy to other sites around the world

    intellectual property and formulation

    capabilities to help with product life cycle

    management.

    Mayne Pharma International has been grant-

    ed, or applied for, patents that protect its vari-

    ous drug delivery technologies. The in-market

    sales of products developed at the Salisbury,

    Australia facility using its technologies are in

    excess of US$500 million per year.

    Mayne Pharmas drug delivery systems

    include:

    Technology to control drug release

    To enable pulsed release, extended release,

    and delayed release profiles (pellet/bead

    formulations produced using extrusion and

    marumerisation, or spheronisation processes,

    see below). Pellets may be tabletted or encap-

    sulated. This technology is very flexible andit can be adapted to the specific formulation

    needs of a particular drug substance.

    Technology to improve oral bioavailability

    Particularly for insoluble drugs (SUBA

    technology, see below).

    Technology to taste mask liquids and tablets

    To improve palatability and aid swallowing

    (Cleantaste technology, see below).

    TECHNOLOGY TO CONTROL

    DRUG RELEASE

    Pellet (or bead) technology allows a vari-

    ety of different drug delivery profiles to be

    achieved by coating drug and excipient with

    various polymers. The drug cores are gener-

    ally spheroidal in shape and have a diameter

    in the range of 300-1,700 m. Pellets can be

    presented in capsule or tablet dosage forms.

    Two types of process are used to generate

    the spheroidal particles (see diagram):

    The first of these processes, which allows

    drug potencies up to 90%, utilises extru-

    sion and marumerisation to form a drug

    core with a polymer coat.

    The second process is known as spheroni-

    sation, where the drug particles are fixed to

    the outside of a seed core (typically a sugar

    sphere). This process provides a very tight

    size distribution of pellets. Drug potencies

    up to 60% are possible.

    For both of the processes above, the desired

    drug release profile is achieved by coating

    these particles with an appropriate polymer.

    Mayne Pharma International has particular

    expertise in polymer selection and process-

    ing. The company can also work with a widerange of solvent systems.

    SUBA

    SUBATM is a novel technology for enhanc-

    ing the bioavailability of poorly water solu-

    ble drugs utilising a solid dispersion of drug

    in various polymers.

    SUBATM has been shown to double the

    oral bioavailability of itraconazole when

    compared with the innovator product(Sporanox).

    CLEANTASTE

    Cleantaste technology allows a polymer

    coat to be applied to very small particles

    (25-150 m diameter) to improve taste. It

    is also possible to use this technology to

    improve stability or to deliver sustained

    release characteristics. The fine, non-gritty

    texture of product produced by this tech-

    nology lends itself to being used in orally

    dispersible tablet and liquid formulations, as

    well as encapsulated products. Cleantaste

    acetaminophen and ambroxol have been

    commercialised and launched in Australia,

    the US and Japan.

    SERVICES SUMMARY

    Mayne Pharma International can develop and

    manufacture oral and topical formulations for

    clinical trials and commercial supply. Mayne

    Pharma International can provide:

    Tablets (immediate, extended, delayed or

    pulsed release and taste masked)

    Capsules (powder, pellets (beads)

    Liquids and Creams

    COMPANY PROFILE - MAYNE PHARMA INTERNATIONAL

    Pellet technology used for controlled release formulations.

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    15/40

    15Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com

    Placebo formulations can be provided to

    match client specifications or innovator

    product. Packaging and labelling can be

    completed to customer requirements.

    In addition to its drug delivery technologies,

    Mayne Pharma International offers a number

    of specialty services:

    Formulation Development

    Provide solutions to a range of common

    formulation challenges such as poor solubil-

    ity, poor bioavailability, short half life, low

    Cmax, poor powder flow, non-uniform crys-tal size and scale-up issues.

    ABOUT MAYNE PHARMA

    A leading pharmaceutical organisation

    built on a heritage of 160 years of

    industry excellence, Mayne Pharma

    International is a market-driven company

    offering a range of drug delivery technolo-

    gies. Mayne Pharma International offers

    contract development and manufacturing

    company for oral and topical pharmaceuti-

    cal products.

    Mayne Pharma international competes in

    the oral drug delivery, branded, generic and

    value-added API markets. The oral pharma-

    ceutical business at Salisbury, Australia, is a

    GMP facility.

    Annual production capacity:

    2,500 million capsules and tablets

    100 tonnes of bulk product

    16 million units of liquids and creams

    The site is approved by all major regulatory

    authorities:

    FDA: United States

    MHRA: UK

    TGA: Australia

    TPD: Canada

    Mayne Pharma International has generated

    numerous patents in the drug delivery field.

    9 patent families

    48 registered patents

    14 pending applications

    Mayne Pharma International is located at

    Salisbury (Adelaide), South Australia. There

    is 12,000 m2 of manufacturing space on a

    19-hectare site. Mayne Pharma International

    is a wholly owned subsidiary of Mayne

    Pharma Group Ltd, an Australian public

    company listed on the ASX.

    Mayne Pharma International

    PO Box 700SalisburySouth Australia 5108Australia

    T: +61 8 8209 2604F: +61 8 8281 6998E: [email protected]

    www.maynepharma.com

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    16/40

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing16

    A new experimental drug substance shows

    great promise from pre-clinical studies for the

    treatment of a disease which afflicts millions of

    patients worldwide. What is the best strategy for

    testing the drug in man for the first time? This

    is a question that all companies developing new

    drugs face on a regular basis.

    Entering Phase I clinical trials is a key

    milestone in any drug development project and

    to reach this stage as quickly as possible is of

    paramount importance especially for those

    with limited budgets. Of equal importance is to

    ensure that the new drug substance is adminis-

    tered in a form that will give it the best chance

    of success in early clinical assessment. A poor

    choice of formulation strategy can lead to poor

    clinical data which can lead to re-formulation

    and a prolonged Phase I clinical programme, or

    even termination of the project.

    So how do you decide what is the best for-

    mulation for a new drug, assuming at this stage

    that it is intended for oral administration?

    KNOW YOUR DRUG SUBSTANCE

    From preclinical studies, there should be

    sufficient information to be able to define the

    drug according to its water-solubility and per-meability characteristics in accordance with the

    biopharmaceutics classification system (BCS).1

    Also, Lipinskis Rule of Five 2 is a useful tool

    in predicting the oral bioavailability of drug

    molecules based on certain molecular attributes.

    The BCS has proved a useful tool to formu-

    lators for classifying drug substances, but its

    primary purpose is for establishing criteria for

    biowaivers, and alternative developability clas-

    sification systems have recently been proposed.3,4

    How well a drug is absorbed into the blood-

    stream from the gastro-intestinal tract (GIT) is

    governed predominantly by (i) drug solubility in

    the gastric and intestinal fluids and (ii) perme-

    ability through cell lipid bilayers. BCS Class I

    drugs are freely soluble in GIT fluids and perme-

    ate easily through lipid bilayers. These drugs are

    well absorbed when given orally and present the

    easiest task when choosing a formulation strat-

    egy. BCS Class IV drugs on the other hand are

    defined as poorly soluble (in GIT fluids) and per-

    meate poorly across lipid bilayers. Consequently,

    these drugs exhibit poor oral bioavailability and

    pose the formulator the greatest challenge.

    Additional physicochemical and biological fac-

    tors which can challenge formulators are:

    Drug instability:

    during processing or in the formulation (e.g.

    apomorphine)

    in the GIT (e.g. when drug is acid labile, as

    with omeprazole). Narrow absorption window in the intestine

    (e.g. acyclovir, captopril).

    Drug metabolism and/or efflux within the

    intestinal wall (e.g. cyclosporin A).

    Making the right choice of formulation for the first-into-human studies of a product candidate

    is extremely important and has significant time and cost implications for the development

    programme. Here, Robert Harris, PhD, Director, Early Development at Molecular Profiles,

    describes various formulation options available and suggests methods that can be used to select

    the best formulation option for a new orally delivered drug substance.

    FROM POWDER TO PILL:

    A RATIONAL APPROACH TO FORMULATINGFOR FIRST-INTO-MAN STUDIES

    Dr Robert Harris

    Director, Early DevelopmentT: +44 115 871 8883F: +44 115 871 8889E: [email protected]

    Molecular Profiles Ltd

    8 Orchard PlaceNottingham Business Park

    NottinghamNG8 6PXUnited Kingdom

    www.molprofiles.com

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    17/40

    Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com 17

    Drug absorption and metabolism can vary

    between animal species and therefore it is notalways possible to predict the influence of bio-

    logical factors (e.g. pre-systemic metabolism)

    on drug uptake in humans from preclinical

    animal studies.

    DECIDE ON A FORMULATION

    STRATEGY

    For first-into-human studies it is usual to

    administer the drug either as powder-in-bottle

    (for reconstitution prior to administration) or in

    capsules, which offer the greatest flexibility for

    dose adjustment. Choosing a formulation will

    depend on the properties of the drug substance

    and the target dose. Decision trees can be very

    effective tools in helping select the most appro-

    priate formulation strategy.5,6 Figure 1 is an

    example of a decision tree which can be used to

    select a suitable formulation strategy for first-

    into-human clinical trials.

    The simplest formulation strategy is not to

    formulate just administer the drug substance

    with no additional excipients. In this case the

    required quantity of drug active is added directly

    to a container (for reconstitution with a suitable

    liquid prior to ingestion) or to a capsule. This

    approach is widely used within the industry as

    it significantly reduces the time and cost for

    progressing to first-into-man studies. For small

    quantities of units the active is weighed into

    each capsule or bottle by hand. For large quanti-

    ties of capsules or where the required dose is

    < 10 mg, capsule filling can be achieved accu-

    rately by use of specialised precision powder

    dosing equipment (for example, Xcelodose

    (Capsugel, Peapack, NJ, US), as shown in

    Figure 2).The drug-in-capsule/bottle approach is par-

    ticularly suited for BCS Class I compounds,

    which are absorbed easily from the GIT.

    Although there are obvious benefits in adopt-

    ing a drug-in-capsule/bottle approach, it should

    be considered with caution if the compound isnot BCS Class I. If a drug substance does not

    wet easily or if its solubility in water is poor the

    drug may be poorly absorbed from the GIT and

    hence exhibit poor bioavailability. If there is a

    known history of poor or variable absorption in

    animal models then a formulation strategy to

    enhance water-solubility of the drug substance

    should be considered.

    Two basic principles for enhancing water-

    solubility of the drug substance are (i) reduction

    of the particle size of the drug substance and (ii)

    use of solubility-enhancing vehicles.

    Brief descriptions of typical solubility-

    enhancing formulation strategies are given

    below. Regardless of the formulation strategy

    chosen, it is vital to assess drug solubility fol-

    lowing dilution of the test formulations in

    aqueous media. The dissolution test procedures

    used should simulate both gastric and intestinal

    conditions (in terms of pH, fluid volume, etc).

    Particle size reduction

    Increasing the overall surface area of a solidcan lead to more rapid dissolution of the drug sub-

    stance. Micronising equipment (e.g. fluid energy

    mills) can reduce particle size down to 2-10

    m. Taking the principle of size reduction even

    further, there are now technologies available to

    produce submicron nanocrystals through pre-

    cipitation (bottom up) or wet milling (top down)

    techniques.7,8 Following particle size reduction

    the drug substance can be dispensed into capsules,

    either as drug alone or as a powder blend (with

    excipients), depending on the required dose and

    flow properties of the milled drug substance.

    Solubility-enhancing vehicles

    For each of the strategies described below

    the resulting formulation can be filled into

    capsule shells for administration. Capsule fill-

    ing machines which are suitable for this pur-

    pose include the IN-CAP (Dott. Bonapace,

    Limbiate, Italy), suitable for powders or liq-

    uids/semi-solids, and the CFS 1200 (Capsugel)

    which is suitable for liquids/semi-solids.

    Solution/semi-solid capsule formulations:

    If the drug can be dissolved in a suitable

    pharmaceutically acceptable vehicle then it may

    be appropriate to consider preparation of a solu-

    tion of the drug which can be filled into cap-

    sules. The main benefit of this approach is that

    pre-dissolving the compound overcomes the

    initial rate limiting step of particulate dissolu-

    tion in the aqueous environment within the GIT.

    However, a potential problem is that the

    drug may precipitate out of solution when

    the formulation disperses in the GIT, par-

    ticularly if the solvent is miscible with water

    (e.g. polyethylene glycol). If the drug is suf-ficiently lipophilic to dissolve in a lipid vehicle

    there is less potential for precipitation on dilu-

    tion in the GIT, as partitioning kinetics will

    favour the drug remaining in the lipid drop-

    Figure 1: Formulation strategy decision tree for first-into-human studies.

    Figure 2: Xcelodose precision powderdispenser.

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    18/40

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing18

    lets. Also, lipidic vehicles are generally well

    absorbed from the GIT and in many cases this

    approach alone can significantly improve the

    oral bioavailability 9,10 compared with admin-

    istration of the solid drug substance, but there

    may be significant inter and intra-subject vari-

    ation in drug uptake, depending on the capac-

    ity of individuals to digest these lipid-based

    formulations.

    In recent years there have been significant

    advances in the use of lipidic excipients and sur-

    factants to produce self-emulsifying drug deliv-

    ery systems (SEDDS) and self-micro-emulsi-

    fying drug delivery systems (SMEDDS) for

    oral drug delivery.11 These formulations form

    emulsions or micro-emulsions spontaneously

    on contact with aqueous media. Both SEDDS

    and SMEDDS use pharmaceutically acceptable

    surfactant excipients to achieve self-emulsifica-

    tion, therefore eliminating the reliance on the

    gastro-intestinal secretions (such as bile salts) to

    emulsify the lipids in the formulation.

    Solid solutions

    Solid solutions12

    (also sometimes describedas solid dispersions) are molecular dispersions

    of the drug molecules in a polymer matrix. This

    approach combines two principles to enhance

    water solubility of a drug:

    1. Conversion of the drug material into its amor-

    phous state generally, a drug substance is

    easier to dissolve when in the amorphous

    state compared with the crystalline state, due

    to absence of ordered intermolecular bonds

    2.Incorporation of the amorphous drug sub-

    stance in a hydrophilic polymeric matrix a

    number of hydrophilic, polymeric materi-

    als have been used as solubility-enhancing

    matrices for drug substances. For example,

    polyvinyl pyrrolidone (PVP) and polyethyl-

    ene glycol (PEG 6000) have been used for

    preparing solid solutions containing poorly

    soluble drugs.

    Solid solutions can be prepared by dis-

    solving both the drug compound and the

    polymer in a suitable volatile solvent. On

    removing the solvent (e.g. by spray drying)

    an amorphous drug-polymer complex is pro-

    duced. On cooling, the drug is then trapped in

    an amorphous state within the water-soluble

    polymer matrix, thus enhancing the water-

    solubility of the drug.

    One potential problem with this type offormulation is that the drug may favour a more

    thermodynamically stable crystalline state,

    which can result in the drug compound crys-

    tallising in the polymer matrix. Therefore the

    physical stability of such formulations needs

    to be assessed using techniques such as differ-

    ential scanning calorimetry (DSC) and X-ray

    crystallography.

    For formulations in which the drug is to be

    dissolved (in liquid or solid vehicles) miscibil-

    ity of the drug substance with the vehicle is

    a key requirement to maximise water-solu-

    bility of the drug and to maintain the physical

    stability of the formulation (i.e. prevent drug

    precipitation). A comparison of the solubility

    parameters for drug and excipients can be used

    to predict miscibility of the drug the excipi-

    ents.13,14,15 The closer together the solubility

    parameters are between drug and excipient the

    higher the probability of the drug and excipi-

    ent being miscible. An example of how this

    information can be used to gauge miscibility

    of drug with excipients is illustrated in Figure3. The graph shows that the polymer with the

    closest spatial proximity to acetaminophen is

    HPMC and we would therefore expect there

    to be a high probability that the drug will be

    miscible in this polymer.

    SOLID DISPERSIONS

    Solid dispersions are similar to solid solu-

    tion formulations, except that the drug exists in

    the form of discrete particles dispersed within a

    polymer or wax matrix.

    MELT EXTRUSION

    This technique 16,17 is an extension of the

    solid solution approach described previously.

    It consists of extruding a co-melt of the drug

    substance and a polymer through a heated screw

    to produce a solid extrudate which can then be

    milled to produce granules (for encapsulation

    or compression into tablets). As with the solid

    solution approach, the production of a melt

    extruded drug/polymer matrix is an effective

    method of increasing the water solubility of a

    poorly water-soluble drug substance. The effec-

    tiveness of this approach depends on miscibility

    of drug and polymer substances and on the drug

    substance and the polymer exhibiting similar

    melting points.

    MELT GRANULATION

    With this approach a water soluble polymer

    is used as a binding agent in a powder mixture

    to produce a granule blend. The blend is heated

    to a temperature at which the polymer bind-ing agent softens (without completely melting)

    which results in formation of aggregates com-

    prised of the drug and excipients. The granule

    mass is then cooled, sieved and is then suitable

    Figure 3: Comparison of acetaminophen and polymer excipients according to theirHansen partial solubility parameters.

    PVP= polyvinyl pyrrolidone PEG= polyethylene glycol EC= ethyl celluloseHEC= hydroxyethyl cellulose PEO= polyethylene oxide HPMC= hydroxypropylmethyl cellulose

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    19/40

    Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com 19

    for either encapsulation or compression into tab-

    lets. This technique has proved to be effective in

    enhancing water-solubility of several drugs.18,19

    INCLUSION COMPLEXES SUCH AS

    CYCLODEXTRINS

    Cyclodextrins 20 are doughnut-shaped mole-

    cules with a lipophilic surface on the inside ring

    and a hydrophilic surface on the outer surface

    of the ring. The principle behind this strategy is

    that the poorly soluble drug molecule fits into

    the inner ring and the outer hydrophilic surface

    of the cyclodextrin holds the complex in solu-

    tion. The inclusion complex can be prepared

    by dissolving the drug and cyclodextrin in a

    common solvent or by solid-state mixing of the

    materials using a high-attrition technique, such

    as ball milling.

    CONCLUSION

    In conclusion, a number of factors need to

    be taken into consideration in deciding how

    best to take a new drug entity into first-into-man

    studies. The drug-in-capsule approach is often

    seen as a cost effective and time saving option

    for testing a drug in Phase I studies. Indeed, it

    significantly reduces the complexity of early

    stage development and progression from drug

    substance to a Phase I clinical trial can be

    achieved within weeks. However, if the drug

    substance has known solubility/bioavailability

    limitations (as is the case for more than 40% of

    NCEs) then due consideration should be given

    to formulation strategies which can enhance

    drug solubility in the GIT.

    Developing a suitable drug formulation

    for first-into-human studies can be prob-

    lematic and time consuming, especially for

    poorly water-soluble drugs. By predicting

    drug-excipient miscibility (through compari-

    son of solubility parameters) and subsequently

    using a decision tree approach for choosing an

    appropriate formulation strategy, it is possible

    to eliminate a significant proportion of trial

    and error from a drug formulation develop-

    ment project. This rational approach to formu-

    lation development offers obvious advantages

    in reducing time for project completion and

    maximising the effectiveness of formulations

    for Phase I studies.

    REFERENCES

    1. FDA Guidance for Industry. Waiver of in

    vivo bioavailability and bioequivalence stud-

    ies for immediate-release solid oral dosage

    forms based on a biopharmaceutics classifi-

    cation system. (August, 2000).

    2. Lipinski, CA et al. Experimental and com-

    putational approaches to estimate solubility

    and permeability in drug discovery and

    development settings. Adv. Drug Deliv.

    Rev. (1997), 23: 3-25.

    3. Wu, CY & Benet, LZ. Predicting drug dis-position via application of BCS: transport/

    absorption/elimination interplay and devel-

    opment of a biopharmaceutics drug disposi-

    tion classification system. Pharm. Res.

    (2005), 22: 11-23.

    4. Butler, JM and Dressman, JB. The devel-

    opability classification system: application

    of biopharmaceutics concepts to formulation

    development. J. Pharm. Sci. (2010), 99:

    4940-4954.

    5. Brachu. S, et al. A decision-support tool

    for the formulation of orally active, poorly

    soluble compounds. Eur. J. Pharm. Sci.

    (2007), 32: 128-139.

    6. Hariharan, M, et al. Reducing the time to

    develop and manufacture formulations for

    first oral dose in humans. Pharm. Tech.,

    October 2003, 68-84.

    7. Kesisoglou, F, et al. Nanosizing Oral

    formulation development and biopharma-

    ceutical evaluation. Adv. Drug Deliv. Rev.

    (2007), 59: 631644.

    8. Eerdenbrugh, BV, et al. Top-down produc-

    tion of drug nanocrystals: Nanosuspension

    stabilisation, miniturization and transforma-

    tion into solid products. Int. J. Pharm.

    (2008), 364: 64-75.

    9. Hauss, DJ. Oral lipid-based formulations.

    Adv. Drug Deliv. Rev. (2007), 59: 667676.

    10. ODriscoll, CM & Griffin, BT.

    Biopharmaceutical challenges associated

    with drugs with low aqueous solubility

    the potential impact of lipid-based formula-

    tions. Adv. Drug Deliv. Rev. (2008), 60:

    617624.

    11. Pouton, CW & Porter, CJH. Formulation

    of lipid-based delivery systems for oral

    administration: Materials, methods and

    strategies. Adv. Drug Deliv. Rev. (2008),

    60: 625637.

    12. Vasconcelos, T, et al. Solid dispersions as

    strategy to improve oral bioavailability of

    poor water soluble drugs. Drug Discovery

    Today (2007), 12: 1068-1075.

    13. Greenhalgh, DJ, et al. Solubility param-

    eters as predictors of miscibility in solid

    dispersions. J. Pharm. Sci. (1999), 88:1182-1190.

    14. Adamska, K, et al. Selection of solubility

    parameters for characterization of phar-

    maceutical excipients. J. Chromatogr. A

    (2007), 1171: 90-97.

    15. Albers, J, et al. Evaluation of predictive

    models for stable solid solution formation.

    J. Pharm. Sci. (2011): 100: 667-680.

    16. Crowley, MM, et al. Pharmaceutical

    applications of hot-melt extrusion: Part

    1. Drug. Dev. Ind. Pharm. (2007), 33:

    909-926

    17. Repka, MA, et al. Pharmaceutical appli-

    cations of hot-melt extrusion: Part 2.

    Drug. Dev. Ind. Pharm. (2007), 33: 1043-

    1057

    18. Yang, D, et al. Effect of the melt granula-

    tion technique on the dissolution charac-

    teristics of griseofulvin. Int. J. Pharm.

    (2007), 329: 72-80.

    19. Passerini, N, et al. Preparation and char-

    acterisation of ibuprofenpoloxamer 188

    granules obtained by melt granulation.

    Eur. J. Pharm. Sci., (2002), 15: 7178.

    20. Brewster, ME & Loftsson, T.

    Cyclodextrins as pharmaceutical solubi-

    lizers. Adv. Drug Deliv. Rev., (2007), 59:

    645666.

    IN WHICH EDITION

    COMPANY APPEAR?WWW.ONDRUGDELIVERY.COM

    COULD YOUR

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    20/40

    www.ondrugdelivery.com Copyright 2011 Frederick Furness Publishing20

    Paediatric and geriatric drug delivery are major

    challenges in drug development: it is estimated

    that 50% of the population have difficulties in

    swallowing solid oral dosage forms. This is espe-

    cially true among children under 12 years and the

    elderly; there is a real need for age-adapted for-

    mulations to promote better treatment compliance.

    Indeed, patients have been found to break

    tablets into fragments in order to facilitate

    administration or to adapt the dose, generating

    major risks such as inaccurate dosing, or stabil-

    ity issues of the residual fragments.

    Liquid formulations are thus one of the most

    appropriate dosage forms for these subpopula-

    tions, as they allow better compliance compared

    with classic tablets or capsules as well as better

    dose adaptability (age- and weight-dependent).

    However, a number of challenges are related

    to the use of liquid formulations:

    The palatability or taste of the solution, which

    must be sufficiently agreeable in flavour to be

    consumed. With respect to bitter-tasting drugs,

    adding sweeteners and flavors to mask the

    taste is often not sufficient.

    A lack of enteric or modified drug delivery tech-

    nologies as compared with tablets and capsules.

    Stability issues of drugs in liquid form.

    LiquiTime, Flamel Technologies innovative

    delivery platform, meets these different challenges.

    Based on a multi-microparticles approach,

    LiquiTime allows stable, controlled-release, ready-

    to-use liquid oral suspensions, with good mouth

    feel, of one or several combined drugs over time.

    The microparticles (shown in Figure 1)

    are composed of a drug core coated with a

    proprietary multifunctional diffusion film. The

    expertise developed by Flamel in coating in

    fluidised beds allows accurate and reproducible

    coating on very small drug cores to manufacture

    microparticles with narrow size distribution and

    final particle diameters below 200 m.

    The microparticles size and the narrow

    distribution optimise mouth-feel, generating a

    smooth, liquid formulation with the possibility

    to adjust the flavour using aroma agents. The

    encapsulation of the active within the micropar-

    ticles allows taste-masking, even for the most

    unpleasant-tasting drugs.

    LiquiTime enables tailoring and accurate fit-

    ting of any release profile, especially zero-order

    kinetics, to optimise pharmacokinetics (Figure 2)

    for a wide range of therapeutic applications and

    drugs (unlike ion exchange resin-complex tech-

    nology, which is limited solely to ionic drugs).

    Other benefits may also be obtained, suchas the possibility to mix immediate-release and

    extended-release kinetics for fast onset and

    extended release, or the possibility of mixing

    different drugs with different release kinetics.

    The multiparticulate nature of the dosage form

    minimises inter- and intra-individual variation as

    compared with conventional tablets or capsules.

    In this article, Camille Rivail, Business Development Analyst, and Jean Chatellier, PhD, Vice-President, Alliance Management, both of Flamel

    Technologies, describe the companys LiquiTime technology, which enables liquid formulations that are palatable, can incorporate variousmodified-release profiles, and are stable with long shelf-lives. The technology meets the need for liquid oral formulations in the large and growing

    number of patients who have difficulty swallowing conventional tablets and capsules, including the young and the elderly.

    LIQUITIME* ORAL LIQUID CONTROLLEDRELEASE DRUG DELIVERY PLATFORM

    Dr Jean Chatellier

    Vice-President, AllianceManagement

    Flamel Technologies SA

    33 avenue du Dr Georges Levy69693 Vnissieux CedexFrance

    T: +33 472 783 434

    F: +33 472 783 446E: [email protected]

    www.flamel.com

    Camille Rivail

    Business Development Analyst

    200m

    Figure 1 : Flamel TechnologiesLiquiTime-based coated microparticleshave an average diameter

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    21/40

    Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com

    Regarding stability, one of the main techni-

    cal hurdles related to liquid forms is to maintain

    the performance of the drug over time to pro-

    vide an acceptable shelf-life. Due to its unique

    approach, LiquiTime has demonstrated long-

    term physical and performance stability of over

    24 months of storage.

    The physical properties of LiquiTime, such as

    viscosity, density, have been optimised to ensure

    precise and reproducible sampling to be delivered

    with existing marketed dosing devices (for exam-

    ple, plastic syringes), allowing flexible and accu-

    rate dose titration adapted to individual patients.

    Development time of LiquiTime formulations

    has been optimised through the use of cutting-edge

    equipment and the skill and experience of the

    Flamel development team. Beyond the lab, cGMP

    manufacturing of clinical trial material and scale-

    up to commercial size can be rapidly executed atFlamels US FDA-approved industrial plant.

    LiquiTime is protected by a strong IP portfo-

    lio, including several granted patents in territo-

    ries including the US, EU and Japan.

    KEY BENEFITS OF LIQUITIME

    Easy to swallow, good mouth feeling, taste

    masked

    Liquid formulations stable over 24 months

    Applicable to a wide range of drugs, not lim-

    ited to ionic drugs as with resin-complex based

    technology

    Zero-order kinetics

    Combination of immediate-release and extend-

    ed-release kinetics possible

    Combination in the same formulation of differ-

    ent drugs with different release kinetics possible

    Use GRAS materials to warrant safety

    Rapid development time under cGMP conditions

    Ease to scale-up to industrial scale

    Clinical Proof of Concept achieved in humans

    for a liquid suspension of an undisclosed drug

    for treatment of children

    Broad and strong IP protection

    ABOUT FLAMEL TECHNOLOGIES

    Flamel Technologies SA (NASDAQ: FLML)

    is a leading drug delivery company focused on the

    goal of developing safer, more efficacious formu-

    lations of drugs that address unmet medical needs.

    Flamel Technologies has collaborations

    with a number of leading pharmaceutical and

    biotechnology companies, including Baxter,

    GlaxoSmithKline (Coreg CR, carvedilol phos-

    phate), Merck Serono and Pfizer.Its product development pipeline includes

    biological and chemical drugs formulated with

    the Micropump, Medusa and other propri-

    etary platforms.

    MICROPUMP

    The Micropump micro-encapsulation oral

    drug delivery platform, for the formulation

    and the controlled release of chemical drugs, is

    designed to increase absorption time, particular-

    ly for drugs only absorbed in the small intestine,

    and to deliver the drug to specific sites in the

    gastro-intestinal tract. Micropump allows tailor-

    ing the exact kinetics required to optimise the

    final product and offers the advantage of easily

    and accurately mixing microparticles with dif-

    ferent release kinetics, in different ratios, with

    every individual particle performing indepen-

    dently. A single Micropump formulation can be

    presented in various dosage forms such as cap-

    sule, tablet, sachet or oral suspensions without

    affecting the release rate.

    Flamel has developed US FDA- and

    EMA-approved products and manufactures

    Micropump-based microparticles.

    TRIGGER LOCK

    In addition to Micropump and LiquiTime,

    Flamel has developed another oral drug delivery

    technology, Trigger Lock, which provides

    controlled release of narcotic and opioid analge-

    sics while deterring tampering (particles cannot

    be crushed to extract the active).

    MEDUSA

    Medusa is a proprietary injectable nanogel

    platform for the formulation and/or the extended

    release of a broad range of biologics (includingproteins, antibodies, peptides and vaccines)

    and of small molecules. The nanogel has been

    proven to be safe and biodegradable (DMF filed

    with the FDA in February 2011).

    Medusa enables the controlled delivery from

    one day up to 14 days of non-modified drugs

    that remain fully active (as opposed to protein

    engineering or chemical modification approach-

    es). It may be used to develop Biobetters with

    potentially improved efficacy, reduced toxic-

    ity and enhanced patient compliance. Several

    Medusa-based products are at various clinical

    stages of development. Flamels lead internal

    Medusa-based product candidate IFN-a XL

    (long-acting interferon alpha-2b) is currently

    the subject of a Phase II trial in HCV patients.

    DeliVax*, Medusas vaccine application,

    permits the efficient formulation of vaccines.

    These versatile drug delivery platforms may

    be used to address threshold formulation prob-

    lems such as poor solubility, aggregation and

    instability for both chemical and biological

    drugs. Flamels innovative delivery platforms

    are used for the lifecycle management of mar-

    keted products, including Biobetters, and the

    development of new compounds with many

    unique competitive advantages:

    Improvement of drug characteristics such as

    efficacy, bioavailability and pharmacokinetics

    Improvement of the drug safety profile with a

    noticeable diminution of peak dose concentra-

    tions, which in turn allows administration of

    higher effective doses and potentially greater

    efficacy

    Potential improvement of patient compliance

    due to reduced side-effects and greater con-

    venience

    Protection of market position through patent

    extension and/or product differentiation Extension of market to new indications and

    new patient populations.

    * pending trademarks

    21

    Figure 2: This graph illustrates the different zero-order release profiles achieved forLiquiTime-based formulations (easily tailored to obtain the appropriated targeted

    product profile).

    Prototype I

    110

    100

    90

    80

    70

    60

    50

    40

    30

    20

    10

    00 2

    Time (hours)

    %c

    umutaliverelease

    d

    4 6 8 10 12 14 16 18 20 22 24

    Prototype II

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    22/40

  • 7/31/2019 Oral Drug Delivery May 2011 Lo Res

    23/40

    Copyright 2011 Frederick Furness Publishing www.ondrugdelivery.com 23

    energy required to remove the volatile liquid

    during the film casting process.

    RELEASE LINERS

    Significant research and expertise derived

    from the transdermal arena has resulted in a

    wide range of release liner technologies that

    may be used as processing aids in the manu-

    facture of OTFs. These materials are comprised

    of a plastic film or paper substrate coated with

    silicone or non-silicone chemistries for a clean

    release of the film when appropriate in the con-

    version process. By coating a compounded liq-

    uid formulation to a continuous web of release

    liner material, film manufacturers are able to

    maintain the integrity of the OTF film product

    throughout the manufacturing process because

    this component provides added strength, sup-

    port and environmental protection to wound

    rolls of OTF film prior to finishing. Release lin-

    ers can be incorporated strictly as a processing

    aide that is removed in the film finishing stage,

    or as seen in new product launches, this com-

    ponent can remain affixed to the OTF to aid in

    dispensing and administering the drug product.

    ACTIVE INGREDIENTS

    OTFs can integrate most available forms of

    APIs, including micronised, granulated, salt,

    and free-base forms. Both soluble and insoluble

    drugs have been successfully compounded into

    solutions, emulsions, or dispersions that have

    subsequently resulted in the launches of the OTFproducts currently available in the market today.

    Larger particle size compounds do present some

    constraints in regards to the final OTFs thick-

    ness, but in general, most APIs and nutritional

    compound particle size distributions fall